43 results on '"Werder, Rhiannon B."'
Search Results
2. Thyroid hormone receptor beta (THRβ1) is the major regulator of T3 action in human iPSC-derived hepatocytes
- Author
-
Soares De Oliveira, Lorraine, Kaserman, Joseph E., Van Der Spek, Anne H., Lee, Nora J., Undeutsch, Hendrik J., Werder, Rhiannon B., Wilson, Andrew A., and Hollenberg, Anthony N.
- Published
- 2024
- Full Text
- View/download PDF
3. Identification of a distal enhancer regulating hedgehog interacting protein gene in human lung epithelial cells
- Author
-
Guo, Feng, Zhang, Li, Yu, Yuzhen, Gong, Lu, Tao, Shiyue, Werder, Rhiannon B., Mishra, Shreya, Zhou, Yihan, Anamika, Wardatul Jannat, Lao, Taotao, Inuzuka, Hiroyuki, Zhang, Yihan, Pham, Betty, Liu, Tao, Tufenkjian, Tiffany S., Richmond, Bradley W., Wei, Wenyi, Mou, Hongmei, Wilson, Andrew A., Hu, Ming, Chen, Wei, and Zhou, Xiaobo
- Published
- 2024
- Full Text
- View/download PDF
4. The COPD GWAS gene ADGRG6 instructs function and injury response in human iPSC-derived type II alveolar epithelial cells
- Author
-
Werder, Rhiannon B., Berthiaume, Kayleigh A., Merritt, Carly, Gallagher, Marissa, Villacorta-Martin, Carlos, Wang, Feiya, Bawa, Pushpinder, Malik, Vidhi, Lyons, Shawn M., Basil, Maria C., Morrisey, Edward E., Kotton, Darrell N., Zhou, Xiaobo, Cho, Michael H., and Wilson, Andrew A.
- Published
- 2023
- Full Text
- View/download PDF
5. Maternal diet modulates the infant microbiome and intestinal Flt3L necessary for dendritic cell development and immunity to respiratory infection
- Author
-
Sikder, Md. Al Amin, Rashid, Ridwan B., Ahmed, Tufael, Sebina, Ismail, Howard, Daniel R., Ullah, Md. Ashik, Rahman, Muhammed Mahfuzur, Lynch, Jason P., Curren, Bodie, Werder, Rhiannon B., Simpson, Jennifer, Bissell, Alec, Morrison, Mark, Walpole, Carina, Radford, Kristen J., Kumar, Vinod, Woodruff, Trent M., Ying, Tan Hui, Ali, Ayesha, Kaiko, Gerard E., Upham, John W., Hoelzle, Robert D., Cuív, Páraic Ó., Holt, Patrick G., Dennis, Paul G., and Phipps, Simon
- Published
- 2023
- Full Text
- View/download PDF
6. Human iPSC-hepatocyte modeling of alpha-1 antitrypsin heterozygosity reveals metabolic dysregulation and cellular heterogeneity
- Author
-
Kaserman, Joseph E., Werder, Rhiannon B., Wang, Feiya, Matte, Taylor, Higgins, Michelle I., Dodge, Mark, Lindstrom-Vautrin, Jonathan, Bawa, Pushpinder, Hinds, Anne, Bullitt, Esther, Caballero, Ignacio S., Shi, Xu, Gerszten, Robert E., Brunetti-Pierri, Nicola, Liesa, Marc, Villacorta-Martin, Carlos, Hollenberg, Anthony N., Kotton, Darrell N., and Wilson, Andrew A.
- Published
- 2022
- Full Text
- View/download PDF
7. Characterization of the COPD alveolar niche using single-cell RNA sequencing
- Author
-
Sauler, Maor, McDonough, John E., Adams, Taylor S., Kothapalli, Neeharika, Barnthaler, Thomas, Werder, Rhiannon B., Schupp, Jonas C., Nouws, Jessica, Robertson, Matthew J., Coarfa, Cristian, Yang, Tao, Chioccioli, Maurizio, Omote, Norihito, Cosme, Jr, Carlos, Poli, Sergio, Ayaub, Ehab A., Chu, Sarah G., Jensen, Klaus H., Gomez, Jose L., Britto, Clemente J., Raredon, Micha Sam B., Niklason, Laura E., Wilson, Andrew A., Timshel, Pascal N., Kaminski, Naftali, and Rosas, Ivan O.
- Published
- 2022
- Full Text
- View/download PDF
8. Adenine base editing reduces misfolded protein accumulation and toxicity in alpha-1 antitrypsin deficient patient iPSC-hepatocytes
- Author
-
Werder, Rhiannon B., Kaserman, Joseph E., Packer, Michael S., Lindstrom-Vautrin, Jonathan, Villacorta-Martin, Carlos, Young, Lauren E., Aratyn-Schaus, Yvonne, Gregoire, Francine, and Wilson, Andrew A.
- Published
- 2021
- Full Text
- View/download PDF
9. Rapid Implementation of a SARS-CoV-2 Diagnostic Quantitative Real-Time PCR Test with Emergency Use Authorization at a Large Academic Safety Net Hospital
- Author
-
Vanuytsel, Kim, Mithal, Aditya, Giadone, Richard M., Yeung, Anthony K., Matte, Taylor M., Dowrey, Todd W., Werder, Rhiannon B., Miller, Gregory J., Miller, Nancy S., Andry, Christopher D., and Murphy, George J.
- Published
- 2020
- Full Text
- View/download PDF
10. Actionable Cytopathogenic Host Responses of Human Alveolar Type 2 Cells to SARS-CoV-2
- Author
-
Hekman, Ryan M., Hume, Adam J., Goel, Raghuveera Kumar, Abo, Kristine M., Huang, Jessie, Blum, Benjamin C., Werder, Rhiannon B., Suder, Ellen L., Paul, Indranil, Phanse, Sadhna, Youssef, Ahmed, Alysandratos, Konstantinos D., Padhorny, Dzmitry, Ojha, Sandeep, Mora-Martin, Alexandra, Kretov, Dmitry, Ash, Peter E.A., Verma, Mamta, Zhao, Jian, Patten, J.J., Villacorta-Martin, Carlos, Bolzan, Dante, Perea-Resa, Carlos, Bullitt, Esther, Hinds, Anne, Tilston-Lunel, Andrew, Varelas, Xaralabos, Farhangmehr, Shaghayegh, Braunschweig, Ulrich, Kwan, Julian H., McComb, Mark, Basu, Avik, Saeed, Mohsan, Perissi, Valentina, Burks, Eric J., Layne, Matthew D., Connor, John H., Davey, Robert, Cheng, Ji-Xin, Wolozin, Benjamin L., Blencowe, Benjamin J., Wuchty, Stefan, Lyons, Shawn M., Kozakov, Dima, Cifuentes, Daniel, Blower, Michael, Kotton, Darrell N., Wilson, Andrew A., Mühlberger, Elke, and Emili, Andrew
- Published
- 2020
- Full Text
- View/download PDF
11. SARS-CoV-2 Infection of Pluripotent Stem Cell-Derived Human Lung Alveolar Type 2 Cells Elicits a Rapid Epithelial-Intrinsic Inflammatory Response
- Author
-
Huang, Jessie, Hume, Adam J., Abo, Kristine M., Werder, Rhiannon B., Villacorta-Martin, Carlos, Alysandratos, Konstantinos-Dionysios, Beermann, Mary Lou, Simone-Roach, Chantelle, Lindstrom-Vautrin, Jonathan, Olejnik, Judith, Suder, Ellen L., Bullitt, Esther, Hinds, Anne, Sharma, Arjun, Bosmann, Markus, Wang, Ruobing, Hawkins, Finn, Burks, Eric J., Saeed, Mohsan, Wilson, Andrew A., Mühlberger, Elke, and Kotton, Darrell N.
- Published
- 2020
- Full Text
- View/download PDF
12. Long-lived regulatory T cells generated during severe bronchiolitis in infancy influence later progression to asthma
- Author
-
Lynch, Jason P., Werder, Rhiannon B., Curren, Bodie F., Sikder, Md. Al Amin, Ullah, Ashik, Sebina, Ismail, Rashid, Ridwan B., Zhang, Vivian, Upham, John W., Hill, Geoff R., Steptoe, Raymond J., and Phipps, Simon
- Published
- 2020
- Full Text
- View/download PDF
13. Reconstructed Single-Cell Fate Trajectories Define Lineage Plasticity Windows during Differentiation of Human PSC-Derived Distal Lung Progenitors
- Author
-
Hurley, Killian, Ding, Jun, Villacorta-Martin, Carlos, Herriges, Michael J., Jacob, Anjali, Vedaie, Marall, Alysandratos, Konstantinos D., Sun, Yuliang L., Lin, Chieh, Werder, Rhiannon B., Huang, Jessie, Wilson, Andrew A., Mithal, Aditya, Mostoslavsky, Gustavo, Oglesby, Irene, Caballero, Ignacio S., Guttentag, Susan H., Ahangari, Farida, Kaminski, Naftali, Rodriguez-Fraticelli, Alejo, Camargo, Fernando, Bar-Joseph, Ziv, and Kotton, Darrell N.
- Published
- 2020
- Full Text
- View/download PDF
14. Chronic IL-33 expression predisposes to virus-induced asthma exacerbations by increasing type 2 inflammation and dampening antiviral immunity
- Author
-
Werder, Rhiannon B., Zhang, Vivian, Lynch, Jason P., Snape, Natale, Upham, John W., Spann, Kirsten, and Phipps, Simon
- Published
- 2018
- Full Text
- View/download PDF
15. Gene-based analysis of regulatory variants identifies 4 putative novel asthma risk genes related to nucleotide synthesis and signaling
- Author
-
Ferreira, Manuel A.R., Jansen, Rick, Willemsen, Gonneke, Penninx, Brenda, Bain, Lisa M., Vicente, Cristina T., Revez, Joana A., Matheson, Melanie C., Hui, Jennie, Tung, Joyce Y., Baltic, Svetlana, Le Souëf, Peter, Montgomery, Grant W., Martin, Nicholas G., Robertson, Colin F., James, Alan, Thompson, Philip J., Boomsma, Dorret I., Hopper, John L., Hinds, David A., Werder, Rhiannon B., and Phipps, Simon
- Published
- 2017
- Full Text
- View/download PDF
16. Aeroallergen-induced IL-33 predisposes to respiratory virus–induced asthma by dampening antiviral immunity
- Author
-
Lynch, Jason P., Werder, Rhiannon B., Simpson, Jennifer, Loh, Zhixuan, Zhang, Vivian, Haque, Ashraful, Spann, Kirsten, Sly, Peter D., Mazzone, Stuart B., Upham, John W., and Phipps, Simon
- Published
- 2016
- Full Text
- View/download PDF
17. IRF-3, IRF-7, and IPS-1 Promote Host Defense against Acute Human Metapneumovirus Infection in Neonatal Mice
- Author
-
Spann, Kirsten M., Loh, Zhixuan, Lynch, Jason P., Ullah, Ashik, Zhang, Vivian, Baturcam, Engin, Werder, Rhiannon B., Khajornjiraphan, Natthida, Rudd, Penny, Loo, Yeuh-Ming, Suhrbier, Andreas, Gale, Michael, Jr., Upham, John W., and Phipps, Simon
- Published
- 2014
- Full Text
- View/download PDF
18. CRISPR interference interrogation of COPD GWAS genes reveals the functional significance of desmoplakin in iPSC-derived alveolar epithelial cells.
- Author
-
Werder, Rhiannon B., Tao Liu, Abo, Kristine M., Lindstrom-Vautrin, Jonathan, Villacorta-Martin, Carlos, Huang, Jessie, Hinds, Anne, Boyer, Nathan, Bullitt, Esther, Liesa, Marc, Silverman, Edwin K., Kotton, Darrell N., Cho, Michael H., Xiaobo Zhou, and Wilson, Andrew A.
- Subjects
- *
EPITHELIAL cells , *PLURIPOTENT stem cells , *OCCLUDINS , *CRISPRS , *MONOCARBOXYLATE transporters , *TRANSFORMING growth factors-beta , *HUMAN biology , *GENOME-wide association studies - Abstract
The article focuses on determining the function of nine genes implicated in COPD by GWAS in induced pluripotent stem cell–derived type 2 alveolar epithelial cells (iAT2s) by applying CRISPR. Topics discussed include biology of potential genetic contributors in the lung to COPD, including genome-wide association studies (GWAS) genes, and CRISPRi-mediated knockdown of COPD GWAS genes alters the iAT2 transcriptional program.
- Published
- 2022
- Full Text
- View/download PDF
19. Human airway lineages derived from pluripotent stem cells reveal the epithelial responses to SARS-CoV-2 infection.
- Author
-
Ruobing Wang, Hume, Adam J., Beermann, Mary Lou, Simone-Roach, Chantelle, Lindstrom-Vautrin, Jonathan, Le Suer, Jake, Huang, Jessie, Olejnik, Judith, Villacorta-Martin, Carlos, Bullitt, Esther, Hinds, Anne, Ghaedi, Mahboobe, Rollins, Stuart, Werder, Rhiannon B., Abo, Kristine M., Wilson, Andrew A., Mühlberger, Elke, Kotton, Darrell N., and Hawkins, Finn J.
- Subjects
PLURIPOTENT stem cells ,INDUCED pluripotent stem cells ,EPITHELIAL cells ,SARS-CoV-2 ,ANGIOTENSIN converting enzyme - Abstract
There is an urgent need to understand how SARS-CoV-2 infects the airway epithelium and in a subset of individuals leads to severe illness or death. Induced pluripotent stem cells (iPSCs) provide a near limitless supply of human cells that can be differentiated into cell types of interest, including airway epithelium, for disease modeling. We present a human iPSC-derived airway epithelial platform, composed of the major airway epithelial cell types, that is permissive to SARS-CoV-2 infection. Subsets of iPSC-airway cells express the SARS-CoV-2 entry factors angiotensin-converting enzyme 2 (ACE2), and transmembrane protease serine 2 (TMPRSS2). Multiciliated cells are the primary initial target of SARS-CoV-2 infection. On infection with SARS-CoV-2, iPSC-airway cells generate robust interferon and inflammatory responses, and treatment with remdesivir or camostat mesylate causes a decrease in viral propagation and entry, respectively. In conclusion, iPSC-derived airway cells provide a physiologically relevant in vitro model system to interrogate the pathogenesis of, and develop treatment strategies for, COVID-19 pneumonia. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
20. Targeting the P2Y13 Receptor Suppresses IL-33 and HMGB1 Release and Ameliorates Experimental Asthma.
- Author
-
Werder, Rhiannon B., Ullah, Md Ashik, Rahman, Muhammed Mahfuzur, Simpson, Jennifer, Lynch, Jason P., Collinson, Natasha, Rittchen, Sonja, Rashid, Ridwan B., Sikder, Md Al Amin, Handoko, Herlina Y., Curren, Bodie F., Sebina, Ismail, Hartel, Gunter, Bissell, Alec, Ngo, Sylvia, Yarlagadda, Tejasri, Hasnain, Sumaira Z., Wenying Lu, Sohal, Sukhwinder S., and Martin, Megan
- Abstract
Rationale: The alarmins IL-33 and HMGB1 (high mobility group box 1) contribute to type 2 inflammation and asthma pathogenesis. Objectives: To determine whether P2Y13-R (P2Y13 receptor), a purinergic GPCR (G protein-coupled receptor) and risk allele for asthma, regulates the release of IL-33 and HMGB1. Methods: Bronchial biopsy specimens were obtained from healthy subjects and subjects with asthma. Primary human airway epithelial cells (AECs), primary mouse AECs, or C57Bl/6 mice were inoculated with various aeroallergens or respiratory viruses, and the nuclear-to-cytoplasmic translocation and release of alarmins was measured by using immunohistochemistry and an ELISA. The role of P2Y13-R in AEC function and in the onset, progression, and exacerbation of experimental asthma was assessed by using pharmacological antagonists and mice with P2Y13-R gene deletion. Measurements and Main Results: Aeroallergen exposure induced the extracellular release of ADP and ATP, nucleotides that activate P2Y13-R. ATP, ADP, and aeroallergen (house dust mite, cockroach, or Alternaria antigen) or virus exposure induced the nuclear-to-cytoplasmic translocation and subsequent release of IL-33 and HMGB1, and this response was ablated by genetic deletion or pharmacological antagonism of P2Y13. In mice, prophylactic or therapeutic P2Y13-R blockade attenuated asthma onset and, critically, ablated the severity of a rhinovirus-associated exacerbation in a high-fidelity experimental model of chronic asthma. Moreover, P2Y13-R antagonism derepressed antiviral immunity, increasing IFN-λ production and decreasing viral copies in the lung. Conclusions: We identify P2Y13-R as a novel gatekeeper of the nuclear alarmins IL-33 and HMGB1 and demonstrate that the targeting of this GPCR via genetic deletion or treatment with a small-molecule antagonist protects against the onset and exacerbations of experimental asthma. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
21. HMGB1 amplifies ILC2-induced type-2 inflammation and airway smooth muscle remodelling.
- Author
-
Loh, Zhixuan, Simpson, Jennifer, Ullah, Ashik, Zhang, Vivian, Gan, Wan J., Lynch, Jason P., Werder, Rhiannon B., Sikder, Al Amin, Lane, Katie, Sim, Choon Boon, Porrello, Enzo, Mazzone, Stuart B., Sly, Peter D., Steptoe, Raymond J., Spann, Kirsten M., Sukkar, Maria B., Upham, John W., and Phipps, Simon
- Subjects
SMOOTH muscle ,ECTOPIC tissue ,RESPIRATORY infections ,INNATE lymphoid cells ,RESPIRATORY organs ,TISSUE remodeling ,HUMAN life cycle - Abstract
Type-2 immunity elicits tissue repair and homeostasis, however dysregulated type-2 responses cause aberrant tissue remodelling, as observed in asthma. Severe respiratory viral infections in infancy predispose to later asthma, however, the processes that mediate tissue damage-induced type-2 inflammation and the origins of airway remodelling remain ill-defined. Here, using a preclinical mouse model of viral bronchiolitis, we find that increased epithelial and mesenchymal high-mobility group box 1 (HMGB1) expression is associated with increased numbers of IL-13-producing type-2 innate lymphoid cell (ILC2s) and the expansion of the airway smooth muscle (ASM) layer. Anti-HMGB1 ablated lung ILC2 numbers and ASM growth in vivo, and inhibited ILC2-mediated ASM cell proliferation in a co-culture model. Furthermore, we identified that HMGB1/RAGE (receptor for advanced glycation endproducts) signalling mediates an ILC2-intrinsic IL-13 auto-amplification loop. In summary, therapeutic targeting of the HMGB1/RAGE signalling axis may act as a novel asthma preventative by dampening ILC2-mediated type-2 inflammation and associated ASM remodelling. Author summary: Asthma can start at any time in life, although most often begins in early childhood. Wheezy viral bronchiolitis is a major independent risk factor for subsequent asthma. However, key knowledge gaps exist in relation to the sequelae of severe viral bronchiolitis and the pathogenic processes that promote type-2 inflammation and airway wall remodelling, cardinal features of asthma. Our study addresses this gap by identifying high-mobility group box 1 as a pathogenic cytokine that contributes to group 2 innate lymphoid cell-induced airway smooth muscle growth. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. Respiratory Syncytial Virus Infection Promotes Necroptosis and HMGB1 Release by Airway Epithelial Cells.
- Author
-
Simpson, Jennifer, Zhixuan Loh, Ullah, Md Ashik, Lynch, Jason P., Werder, Rhiannon B., Collinson, Natasha, Zhang, Vivian, Dondelinger, Yves, Bertrand, Mathieu J. M., Everard, Mark L., Blyth, Christopher C., Hartel, Gunter, Van Oosterhout, Antoon J., Gough, Peter J., Bertin, John, Upham, John W., Spann, Kirsten M., Phipps, Simon, and Loh, Zhixuan
- Subjects
RESPIRATORY syncytial virus infections ,HIGH mobility group proteins ,EPITHELIAL cells ,BRONCHIOLITIS ,DISEASE progression ,PROTEIN metabolism ,RESPIRATORY mucosa ,RESEARCH ,ANIMAL experimentation ,RESEARCH methodology ,EVALUATION research ,MEDICAL cooperation ,COMPARATIVE studies ,BRONCHIOLE diseases ,QUESTIONNAIRES ,MICE ,LONGITUDINAL method - Abstract
Rationale: Respiratory syncytial virus (RSV) bronchiolitis causes significant infant mortality. Bronchiolitis is characterized by airway epithelial cell (AEC) death; however, the mode of death remains unknown.Objectives: To determine whether necroptosis contributes to RSV bronchiolitis pathogenesis via HMGB1 (high mobility group box 1) release.Methods: Nasopharyngeal samples were collected from children presenting to the hospital with acute respiratory infection. Primary human AECs and neonatal mice were inoculated with RSV and murine Pneumovirus, respectively. Necroptosis was determined via viability assays and immunohistochemistry for RIPK1 (receptor-interacting protein kinase-1), MLKL (mixed lineage kinase domain-like pseudokinase) protein, and caspase-3. Necroptosis was blocked using pharmacological inhibitors and RIPK1 kinase-dead knockin mice.Measurements and Main Results: HMGB1 levels were elevated in nasopharyngeal samples of children with acute RSV infection. RSV-induced epithelial cell death was associated with increased phosphorylated RIPK1 and phosphorylated MLKL but not active caspase-3 expression. Inhibition of RIPK1 or MLKL attenuated RSV-induced HMGB1 translocation and release, and lowered viral load. MLKL inhibition increased active caspase-3 expression in a caspase-8/9-dependent manner. In susceptible mice, Pneumovirus infection upregulated RIPK1 and MLKL expression in the airway epithelium at 8 to 10 days after infection, coinciding with AEC sloughing, HMGB1 release, and neutrophilic inflammation. Genetic or pharmacological inhibition of RIPK1 or MLKL attenuated these pathologies, lowered viral load, and prevented type 2 inflammation and airway remodeling. Necroptosis inhibition in early life ameliorated asthma progression induced by viral or allergen challenge in later life.Conclusions: Pneumovirus infection induces AEC necroptosis. Inhibition of necroptosis may be a viable strategy to limit the severity of viral bronchiolitis and break its nexus with asthma. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
23. Actionable Cytopathogenic Host Responses of Human Alveolar Type 2 Cells to SARS-CoV-2
- Author
-
Hekman, Ryan M., Hume, Adam J., Goel, Raghuveera Kumar, Abo, Kristine M., Huang, Jessie, Blum, Benjamin C., Werder, Rhiannon B., Suder, Ellen L., Paul, Indranil, Phanse, Sadhna, Youssef, Ahmed, Alysandratos, Konstantinos D., Padhorny, Dzmitry, Ojha, Sandeep, Mora-Martin, Alexandra, Kretov, Dmitry, Ash, Peter E.A., Verma, Mamta, Zhao, Jian, Patten, J.J., Villacorta-Martin, Carlos, Bolzan, Dante, Perea-Resa, Carlos, Bullitt, Esther, Hinds, Anne, Tilston-Lunel, Andrew, Varelas, Xaralabos, Farhangmehr, Shaghayegh, Braunschweig, Ulrich, Kwan, Julian H., McComb, Mark, Basu, Avik, Saeed, Mohsan, Perissi, Valentina, Burks, Eric J., Layne, Matthew D., Connor, John H., Davey, Robert, Cheng, Ji-Xin, Wolozin, Benjamin L., Blencowe, Benjamin J., Wuchty, Stefan, Lyons, Shawn M., Kozakov, Dima, Cifuentes, Daniel, Blower, Michael, Kotton, Darrell N., Wilson, Andrew A., Mühlberger, Elke, and Emili, Andrew
- Published
- 2021
- Full Text
- View/download PDF
24. qRT-PCR Platforms for Diagnosing and Reporting SARS-CoV-2 Infection in Human Samples
- Author
-
Giadone, Richard M., Mithal, Aditya, Miller, Gregory J., Matte, Taylor M., Yeung, Anthony K., Dowrey, Todd W., Werder, Rhiannon B., Miller, Nancy S., Andry, Christopher D., Vanuytsel, Kim, and Murphy, George J.
- Published
- 2020
- Full Text
- View/download PDF
25. Gene-based analysis of regulatory variants identifies four putative novel asthma risk genes related to nucleotide synthesis and signaling
- Author
-
Ferreira, Manuel A.R., Jansen, Rick, Willemsen, Gonneke, Penninx, Brenda, Bain, Lisa M, Vicente, Cristina T, Revez, Joana A, Matheson, Melanie C., Hui, Jennie, Tung, Joyce Y., Baltic, Svetlana, Le Souëf, Peter, Montgomery, Grant W., Martin, Nicholas G., Robertson, Colin F., James, Alan, Thompson, Philip J., Boomsma, Dorret I., Hopper, John L., Hinds, David A., Werder, Rhiannon B., and Phipps, Simon
- Subjects
Nucleotides ,Receptors, Purinergic P2 ,Quantitative Trait Loci ,Genetic Variation ,Mitochondrial Proton-Translocating ATPases ,Article ,Asthma ,Mice, Inbred C57BL ,Mice ,Receptors, Purinergic P2Y ,Animals ,Humans ,Genetic Predisposition to Disease ,Software ,Genome-Wide Association Study - Abstract
Hundreds of genetic variants are thought to contribute to variation in asthma risk by modulating gene expression. Methods that increase the power of genome-wide association studies (GWASs) to identify risk-associated variants are needed.We sought to develop a method that aggregates the evidence for association with disease risk across expression quantitative trait loci (eQTLs) of a gene and use this approach to identify asthma risk genes.We developed a gene-based test and software package called EUGENE that (1) is applicable to GWAS summary statistics; (2) considers both cis- and trans-eQTLs; (3) incorporates eQTLs identified in different tissues; and (4) uses simulations to account for multiple testing. We applied this approach to 2 published asthma GWASs (combined n = 46,044) and used mouse studies to provide initial functional insights into 2 genes with novel genetic associations.We tested the association between asthma and 17,190 genes that were found to have cis- and/or trans-eQTLs across 16 published eQTL studies. At an empirical FDR of 5%, 48 genes were associated with asthma risk. Of these, for 37, the association was driven by eQTLs located in established risk loci for allergic disease, including 6 genes not previously implicated in disease cause (eg, LIMS1, TINF2, and SAFB). The remaining 11 significant genes represent potential novel genetic associations with asthma. The association with 4 of these replicated in an independent GWAS: B4GALT3, USMG5, P2RY13, and P2RY14, which are genes involved in nucleotide synthesis or nucleotide-dependent cell activation. In mouse studies, P2ry13 and P2ry14-purinergic receptors activated by adenosine 5-diphosphate and UDP-sugars, respectively-were upregulated after allergen challenge, notably in airway epithelial cells, eosinophils, and neutrophils. Intranasal exposure with receptor agonists induced the release of IL-33 and subsequent eosinophil infiltration into the lungs.We identified novel associations between asthma and eQTLs for 4 genes related to nucleotide synthesis/signaling and demonstrated the power of gene-based analyses of GWASs.
- Published
- 2016
26. PGD2/DP2 receptor activation promotes severe viral bronchiolitis by suppressing IFN-1 production.
- Author
-
Werder, Rhiannon B., Lynch, Jason P., Simpson, Jennifer C., Zhang, Vivian, Hodge, Nick H., Poh, Matthew, Forbes-Blom, Elizabeth, Kulis, Christina, Smythe, Mark L., Upham, John W., Spann, Kirsten, Everard, Mark L., and Phipps, Simon
- Subjects
PROSTAGLANDINS ,RESPIRATORY diseases ,RESPIRATORY syncytial virus infections ,ANTINEOPLASTIC agents ,BRONCHIOLITIS ,LYMPHOCYTES ,ASTHMA - Abstract
RSV-induced prostaglandin D2 release contributes to disease severity by impairing IFN-λ production. RSV gives innate immunity the runaround: Asthma can be exacerbated by pathogens such as respiratory syncytial virus (RSV); prostaglandin D2 (PGD2) is also important in asthma and is being investigated as a therapeutic target. Werder et al. used multiple models to examine how RSV infection may perturb immune responses and influence asthma pathogenesis. Samples from infants with bronchiolitis or primary pediatric epithelial cells infected with RSV had elevated PGD2. Modulating PGD2 signaling in a mouse model of severe bronchiolitis improved antiviral immunity and dampened asthmatic symptoms later in life. This protection was not due to preventing type 2 immunity but instead a restoration of IFN-λ production. Their findings shed light on this host-pathogen interaction and suggest new therapeutic avenues. Prostaglandin D2 (PGD2) signals through PGD2 receptor 2 (DP2, also known as CRTH2) on type 2 effector cells to promote asthma pathogenesis; however, little is known about its role during respiratory syncytial virus (RSV) bronchiolitis, a major risk factor for asthma development. We show that RSV infection up-regulated hematopoietic prostaglandin D synthase expression and increased PGD2 release by cultured human primary airway epithelial cells (AECs). Moreover, PGD2 production was elevated in nasopharyngeal samples from young infants hospitalized with RSV bronchiolitis compared to healthy controls. In a neonatal mouse model of severe viral bronchiolitis, DP2 antagonism decreased viral load, immunopathology, and morbidity and ablated the predisposition for subsequent asthma onset in later life. This protective response was abolished upon dual DP1/DP2 antagonism and replicated with a specific DP1 agonist. Rather than mediating an effect via type 2 inflammation, the beneficial effects of DP2 blockade or DP1 agonism were associated with increased interferon-λ (IFN-λ) [interleukin-28A/B (IL-28A/B)] expression and were lost upon IL-28A neutralization. In RSV-infected AEC cultures, DP1 activation up-regulated IFN-λ production, which, in turn, increased IFN-stimulated gene expression, accelerating viral clearance. Our findings suggest that DP2 antagonists or DP1 agonists may be useful antivirals for the treatment of viral bronchiolitis and possibly as primary preventatives for asthma. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
27. The Influence of the Microbiome on Early-Life Severe Viral Lower Respiratory Infections and Asthma -- Food for Thought?
- Author
-
Lynch, Jason P., Al Amin Sikder, Md., Curren, Bodie F., Werder, Rhiannon B., Simpson, Jennifer, Cuív, Páraic Ó., Dennis, Paul G., Everard, Mark L., and Phipps, Simon
- Subjects
RESPIRATORY infections ,ASTHMA ,LUNG microbiology - Abstract
Severe viral lower respiratory infections are a major cause of infant morbidity. In developing countries, respiratory syncytial virus (RSV)-bronchiolitis induces significant mortality, whereas in developed nations the disease represents a major risk factor for subsequent asthma. Susceptibility to severe RSV-bronchiolitis is governed by gene-environmental interactions that affect the host response to RSV infection. Emerging evidence suggests that the excessive inflammatory response and ensuing immunopathology, typically as a consequence of insufficient immunoregulation, leads to long-term changes in immune cells and structural cells that render the host susceptible to subsequent environmental incursions. Thus, the initial host response to RSV may represent a tipping point in the balance between long-term respiratory health or chronic disease (e.g., asthma). The composition and diversity of the microbiota, which in humans stabilizes in the first year of life, critically affects the development and function of the immune system. Hence, perturbations to the maternal and/or infant microbiota are likely to have a profound impact on the host response to RSV and susceptibility to childhood asthma. Here, we review recent insights describing the effects of the microbiota on immune system homeostasis and respiratory disease and discuss the environmental factors that promote microbial dysbiosis in infancy. Ultimately, this knowledge will be harnessed for the prevention and treatment of severe viral bronchiolitis as a strategy to prevent the onset and development of asthma. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
28. RAGE deficiency predisposes mice to virus-induced paucigranulocytic asthma.
- Author
-
Arikkatt, Jaisy, Ullah, Md Ashik, Short, Kirsty Renfree, Vivan Zhang, Wan Jun Gan, Zhixuan Loh, Werder, Rhiannon B., Simpson, Jennifer, Sly, Peter D., Mazzone, Stuart B., Spann, Kirsten M., Ferreira, Manuel A. R., Upham, John W., Sukkar, Maria B., and Phipps, Simon
- Published
- 2017
- Full Text
- View/download PDF
29. 196: Critical role for IL-33 in the onset and progression of experimental asthma induced by concomitant virus infection and allergen exposure
- Author
-
Werder, Rhiannon B., Lynch, Jason P., Simpson, Jennifer, Loh, Ken, Zhang, Vivian, Spann, Kirsten, Sly, Peter D., Mazzone, Stuart, Upham, John W., and Phipps, Simon
- Published
- 2014
- Full Text
- View/download PDF
30. Human iPSC-hepatocyte modeling of alpha-1 antitrypsin heterozygosity reveals metabolic dysregulation and cellular heterogeneity
- Author
-
Joseph E. Kaserman, Rhiannon B. Werder, Feiya Wang, Taylor Matte, Michelle I. Higgins, Mark Dodge, Jonathan Lindstrom-Vautrin, Pushpinder Bawa, Anne Hinds, Esther Bullitt, Ignacio S. Caballero, Xu Shi, Robert E. Gerszten, Nicola Brunetti-Pierri, Marc Liesa, Carlos Villacorta-Martin, Anthony N. Hollenberg, Darrell N. Kotton, Andrew A. Wilson, Kaserman, Joseph E, Werder, Rhiannon B, Wang, Feiya, Matte, Taylor, Higgins, Michelle I, Dodge, Mark, Lindstrom-Vautrin, Jonathan, Bawa, Pushpinder, Hinds, Anne, Bullitt, Esther, Caballero, Ignacio S, Shi, Xu, Gerszten, Robert E, Brunetti-Pierri, Nicola, Liesa, Marc, Villacorta-Martin, Carlo, Hollenberg, Anthony N, Kotton, Darrell N, Wilson, Andrew A, National Health and Medical Research Council (Australia), National Institutes of Health (US), and Alpha- Foundation
- Subjects
alpha-1 antitrypsin deficiency ,metabolic dysregulation ,induced pluripotent stem cell ,Liver fibrosis ,Induced Pluripotent Stem Cells ,liver fibrosi ,unfolded protein response ,proteostasi ,General Biochemistry, Genetics and Molecular Biology ,CP: Metabolism ,CP: Stem cell research ,iPSC-derived hepatocytes ,cellular heterogeneity ,mitochondrial dysfunction ,Proteostasis ,Hepatocytes ,Humans ,ER stre ,ER stress ,iPSC-derived hepatocyte - Abstract
Individuals homozygous for the “Z” mutation in alpha-1 antitrypsin deficiency are known to be at increased risk for liver disease. It has also become clear that some degree of risk is similarly conferred by the heterozygous state. A lack of model systems that recapitulate heterozygosity in human hepatocytes has limited the ability to study the impact of a single Z alpha-1 antitrypsin (ZAAT) allele on hepatocyte biology. Here, we describe the derivation of syngeneic induced pluripotent stem cells (iPSCs) engineered to determine the effects of ZAAT heterozygosity in iPSC-hepatocytes (iHeps). We find that heterozygous MZ iHeps exhibit an intermediate disease phenotype and share with ZZ iHeps alterations in AAT protein processing and downstream perturbations including altered endoplasmic reticulum (ER) and mitochondrial morphology, reduced mitochondrial respiration, and branch-specific activation of the unfolded protein response in cell subpopulations. Our model of MZ heterozygosity thus provides evidence that a single Z allele is sufficient to disrupt hepatocyte homeostatic function., This work was supported by an Alpha-1 Foundation John W. Walsh Translational Research Award (to J.E.K.); a CJ Martin Early Career Fellowship from the Australian National Health and Medical Research Council (to R.B.W.); NIH grant R01HL095993 (to D.N.K.); and NIH grants R01DK101501 (to A.A.W.) and R01DK117940 (to A.N.H. and A.A.W.). iPSC distribution and disease modeling is supported by NIH grants U01TR001810 (to D.N.K. and A.A.W.) and N0175N92020C00005 (to D.N.K.); and by The Alpha-1 Project (TAP), a wholly owned subsidiary of the Alpha-1 Foundation (to D.N.K. and A.A.W.).
- Published
- 2022
31. Breathing new life into the study of COPD with genes identified from genome-wide association studies.
- Author
-
Werder RB, Zhou X, Cho MH, and Wilson AA
- Subjects
- Humans, Risk Factors, Animals, Genetic Markers, Prognosis, Pulmonary Disease, Chronic Obstructive genetics, Pulmonary Disease, Chronic Obstructive physiopathology, Pulmonary Disease, Chronic Obstructive diagnosis, Genome-Wide Association Study, Genetic Predisposition to Disease, Lung physiopathology, Phenotype
- Abstract
COPD is a major cause of morbidity and mortality globally. While the significance of environmental exposures in disease pathogenesis is well established, the functional contribution of genetic factors has only in recent years drawn attention. Notably, many genes associated with COPD risk are also linked with lung function. Because reduced lung function precedes COPD onset, this association is consistent with the possibility that derangements leading to COPD could arise during lung development. In this review, we summarise the role of leading genes ( HHIP , FAM13A , DSP , AGER and TGFB2 ) identified by genome-wide association studies in lung development and COPD. Because many COPD genome-wide association study genes are enriched in lung epithelial cells, we focus on the role of these genes in the lung epithelium in development, homeostasis and injury., Competing Interests: Conflict of interest: X. Zhou has received grant support from Bayer. M.H. Cho has received grant support from GSK and Bayer; and has received speaking/consulting fees from Illumina, AstraZeneca and Genentech. R.B. Werder and A.A. Wilson declared no conflicts of interest., (Copyright ©The authors 2024.)
- Published
- 2024
- Full Text
- View/download PDF
32. Generating 3D Spheres and 2D Air-Liquid Interface Cultures of Human Induced Pluripotent Stem Cell-Derived Type 2 Alveolar Epithelial Cells.
- Author
-
Werder RB, Huang J, Abo KM, Hix OT, Minakin K, Alysandratos KD, Merritt C, Berthiaume K, Alber AB, Burgess CL, Kotton DN, and Wilson AA
- Subjects
- Adult, Alveolar Epithelial Cells, Cell Differentiation, Epithelium, Humans, Induced Pluripotent Stem Cells, Pulmonary Surfactants
- Abstract
In the lung, the alveolar epithelium is a physical barrier from environmental stimuli and plays an essential role in homeostasis and disease. Type 2 alveolar epithelial cells (AT2s) are the facultative progenitors of the distal lung epithelium. Dysfunction and injury of AT2s can result from and contribute to various lung diseases. Improved understanding of AT2 biology is, thus, critical for understanding lung biology and disease; however, primary human AT2s are generally difficult to isolate and limited in supply. To overcome these limitations, human induced pluripotent stem cell (iPSC)-derived type 2 alveolar epithelial cells (iAT2s) can be generated through a directed differentiation protocol that recapitulates in vivo lung development. iAT2s grow in feeder-free conditions, share a transcriptomic program with human adult primary AT2s, and execute key functions of AT2s such as production, packaging, and secretion of surfactant. This protocol details the methods for maintaining self-renewing iAT2s through serial passaging in three-dimensional (3D) culture or adapting iAT2s to air-liquid interface (ALI) culture. A single-cell suspension of iAT2s is generated before plating in 3D solubilized basement membrane matrix (hereafter referred to as "matrix"), where they self-assemble into monolayered epithelial spheres. iAT2s in 3D culture can be serially dissociated into single-cell suspensions to be passaged or plated in 2D ALI culture. In ALI culture, iAT2s form a polarized monolayer with the apical surface exposed to air, making this platform readily amenable to environmental exposures. Hence, this protocol generates an inexhaustible supply of iAT2s, producing upwards of 1 x 10
30 cells per input cell over 15 passages while maintaining the AT2 program indicated by SFTPCtdTomato expression. The resulting cells represent a reproducible and relevant platform that can be applied to study genetic mutations, model environmental exposures, or screen drugs.- Published
- 2022
- Full Text
- View/download PDF
33. Air-liquid interface culture promotes maturation and allows environmental exposure of pluripotent stem cell-derived alveolar epithelium.
- Author
-
Abo KM, Sainz de Aja J, Lindstrom-Vautrin J, Alysandratos KD, Richards A, Garcia-de-Alba C, Huang J, Hix OT, Werder RB, Bullitt E, Hinds A, Falconer I, Villacorta-Martin C, Jaenisch R, Kim CF, Kotton DN, and Wilson AA
- Subjects
- Environmental Exposure, Epithelium, Humans, Electronic Nicotine Delivery Systems, Induced Pluripotent Stem Cells metabolism, Pluripotent Stem Cells
- Abstract
Type 2 alveolar epithelial cells (AT2s), facultative progenitor cells of the lung alveolus, play a vital role in the biology of the distal lung. In vitro model systems that incorporate human cells, recapitulate the biology of primary AT2s, and interface with the outside environment could serve as useful tools to elucidate functional characteristics of AT2s in homeostasis and disease. We and others recently adapted human induced pluripotent stem cell-derived AT2s (iAT2s) for air-liquid interface (ALI) culture. Here, we comprehensively characterize the effects of ALI culture on iAT2s and benchmark their transcriptional profile relative to both freshly sorted and cultured primary human fetal and adult AT2s. We find that iAT2s cultured at ALI maintain an AT2 phenotype while upregulating expression of transcripts associated with AT2 maturation. We then leverage this platform to assay the effects of exposure to clinically significant, inhaled toxicants including cigarette smoke and electronic cigarette vapor.
- Published
- 2022
- Full Text
- View/download PDF
34. Human airway lineages derived from pluripotent stem cells reveal the epithelial responses to SARS-CoV-2 infection.
- Author
-
Wang R, Hume AJ, Beermann ML, Simone-Roach C, Lindstrom-Vautrin J, Le Suer J, Huang J, Olejnik J, Villacorta-Martin C, Bullitt E, Hinds A, Ghaedi M, Rollins S, Werder RB, Abo KM, Wilson AA, Mühlberger E, Kotton DN, and Hawkins FJ
- Subjects
- Epithelial Cells, Humans, SARS-CoV-2, COVID-19, Induced Pluripotent Stem Cells, Pluripotent Stem Cells
- Abstract
There is an urgent need to understand how SARS-CoV-2 infects the airway epithelium and in a subset of individuals leads to severe illness or death. Induced pluripotent stem cells (iPSCs) provide a near limitless supply of human cells that can be differentiated into cell types of interest, including airway epithelium, for disease modeling. We present a human iPSC-derived airway epithelial platform, composed of the major airway epithelial cell types, that is permissive to SARS-CoV-2 infection. Subsets of iPSC-airway cells express the SARS-CoV-2 entry factors angiotensin-converting enzyme 2 ( ACE2 ), and transmembrane protease serine 2 ( TMPRSS2 ). Multiciliated cells are the primary initial target of SARS-CoV-2 infection. On infection with SARS-CoV-2, iPSC-airway cells generate robust interferon and inflammatory responses, and treatment with remdesivir or camostat mesylate causes a decrease in viral propagation and entry, respectively. In conclusion, iPSC-derived airway cells provide a physiologically relevant in vitro model system to interrogate the pathogenesis of, and develop treatment strategies for, COVID-19 pneumonia.
- Published
- 2022
- Full Text
- View/download PDF
35. The oral drug nitazoxanide restricts SARS-CoV-2 infection and attenuates disease pathogenesis in Syrian hamsters.
- Author
-
Miorin L, Mire CE, Ranjbar S, Hume AJ, Huang J, Crossland NA, White KM, Laporte M, Kehrer T, Haridas V, Moreno E, Nambu A, Jangra S, Cupic A, Dejosez M, Abo KA, Tseng AE, Werder RB, Rathnasinghe R, Mutetwa T, Ramos I, de Aja JS, de Alba Rivas CG, Schotsaert M, Corley RB, Falvo JV, Fernandez-Sesma A, Kim C, Rossignol JF, Wilson AA, Zwaka T, Kotton DN, Mühlberger E, García-Sastre A, and Goldfeld AE
- Abstract
A well-tolerated and cost-effective oral drug that blocks SARS-CoV-2 growth and dissemination would be a major advance in the global effort to reduce COVID-19 morbidity and mortality. Here, we show that the oral FDA-approved drug nitazoxanide (NTZ) significantly inhibits SARS-CoV-2 viral replication and infection in different primate and human cell models including stem cell-derived human alveolar epithelial type 2 cells. Furthermore, NTZ synergizes with remdesivir, and it broadly inhibits growth of SARS-CoV-2 variants B.1.351 (beta), P.1 (gamma), and B.1617.2 (delta) and viral syncytia formation driven by their spike proteins. Strikingly, oral NTZ treatment of Syrian hamsters significantly inhibits SARS-CoV-2-driven weight loss, inflammation, and viral dissemination and syncytia formation in the lungs. These studies show that NTZ is a novel host-directed therapeutic that broadly inhibits SARS-CoV-2 dissemination and pathogenesis in human and hamster physiological models, which supports further testing and optimization of NTZ-based therapy for SARS-CoV-2 infection alone and in combination with antiviral drugs.
- Published
- 2022
- Full Text
- View/download PDF
36. Targeting the P2Y 13 Receptor Suppresses IL-33 and HMGB1 Release and Ameliorates Experimental Asthma.
- Author
-
Werder RB, Ullah MA, Rahman MM, Simpson J, Lynch JP, Collinson N, Rittchen S, Rashid RB, Sikder MAA, Handoko HY, Curren BF, Sebina I, Hartel G, Bissell A, Ngo S, Yarlagadda T, Hasnain SZ, Lu W, Sohal SS, Martin M, Bowler S, Burr LD, Martinez LO, Robaye B, Spann K, Ferreira MAR, and Phipps S
- Subjects
- Animals, Asthma metabolism, Asthma physiopathology, Biomarkers metabolism, Case-Control Studies, Disease Progression, Enzyme-Linked Immunosorbent Assay, Epithelial Cells metabolism, Humans, Immunohistochemistry, Mice, Mice, Inbred C57BL, Asthma immunology, HMGB1 Protein metabolism, Interleukin-33 metabolism, Receptors, Purinergic P2 metabolism
- Abstract
Rationale: The alarmins IL-33 and HMGB1 (high mobility group box 1) contribute to type 2 inflammation and asthma pathogenesis. Objectives: To determine whether P2Y
13 -R (P2Y13 receptor), a purinergic GPCR (G protein-coupled receptor) and risk allele for asthma, regulates the release of IL-33 and HMGB1. Methods: Bronchial biopsy specimens were obtained from healthy subjects and subjects with asthma. Primary human airway epithelial cells (AECs), primary mouse AECs, or C57Bl/6 mice were inoculated with various aeroallergens or respiratory viruses, and the nuclear-to-cytoplasmic translocation and release of alarmins was measured by using immunohistochemistry and an ELISA. The role of P2Y13 -R in AEC function and in the onset, progression, and exacerbation of experimental asthma was assessed by using pharmacological antagonists and mice with P2Y13 -R gene deletion. Measurements and Main Results: Aeroallergen exposure induced the extracellular release of ADP and ATP, nucleotides that activate P2Y13 -R. ATP, ADP, and aeroallergen (house dust mite, cockroach, or Alternaria antigen) or virus exposure induced the nuclear-to-cytoplasmic translocation and subsequent release of IL-33 and HMGB1, and this response was ablated by genetic deletion or pharmacological antagonism of P2Y13. In mice, prophylactic or therapeutic P2Y13 -R blockade attenuated asthma onset and, critically, ablated the severity of a rhinovirus-associated exacerbation in a high-fidelity experimental model of chronic asthma. Moreover, P2Y13 -R antagonism derepressed antiviral immunity, increasing IFN-λ production and decreasing viral copies in the lung. Conclusions: We identify P2Y13 -R as a novel gatekeeper of the nuclear alarmins IL-33 and HMGB1 and demonstrate that the targeting of this GPCR via genetic deletion or treatment with a small-molecule antagonist protects against the onset and exacerbations of experimental asthma.- Published
- 2022
- Full Text
- View/download PDF
37. SARS-CoV-2 Infection of Pluripotent Stem Cell-derived Human Lung Alveolar Type 2 Cells Elicits a Rapid Epithelial-Intrinsic Inflammatory Response.
- Author
-
Huang J, Hume AJ, Abo KM, Werder RB, Villacorta-Martin C, Alysandratos KD, Beermann ML, Simone-Roach C, Lindstrom-Vautrin J, Olejnik J, Suder EL, Bullitt E, Hinds A, Sharma A, Bosmann M, Wang R, Hawkins F, Burks EJ, Saeed M, Wilson AA, Mühlberger E, and Kotton DN
- Abstract
The most severe and fatal infections with SARS-CoV-2 result in the acute respiratory distress syndrome, a clinical phenotype of coronavirus disease 2019 (COVID-19) that is associated with virions targeting the epithelium of the distal lung, particularly the facultative progenitors of this tissue, alveolar epithelial type 2 cells (AT2s). Little is known about the initial responses of human lung alveoli to SARS-CoV-2 infection due in part to inability to access these cells from patients, particularly at early stages of disease. Here we present an in vitro human model that simulates the initial apical infection of the distal lung epithelium with SARS-CoV-2, using AT2s that have been adapted to air-liquid interface culture after their derivation from induced pluripotent stem cells (iAT2s). We find that SARS-CoV-2 induces a rapid global transcriptomic change in infected iAT2s characterized by a shift to an inflammatory phenotype predominated by the secretion of cytokines encoded by NF-kB target genes, delayed epithelial interferon responses, and rapid loss of the mature lung alveolar epithelial program. Over time, infected iAT2s exhibit cellular toxicity that can result in the death of these key alveolar facultative progenitors, as is observed in vivo in COVID-19 lung autopsies. Importantly, drug testing using iAT2s confirmed an antiviral dose-response to remdesivir and demonstrated the efficacy of TMPRSS2 protease inhibition, validating a putative mechanism used for viral entry in human alveolar cells. Our model system reveals the cell-intrinsic responses of a key lung target cell to infection, providing a physiologically relevant platform for further drug development and facilitating a deeper understanding of COVID-19 pathogenesis.
- Published
- 2020
- Full Text
- View/download PDF
38. Human iPSC-derived alveolar and airway epithelial cells can be cultured at air-liquid interface and express SARS-CoV-2 host factors.
- Author
-
Abo KM, Ma L, Matte T, Huang J, Alysandratos KD, Werder RB, Mithal A, Beermann ML, Lindstrom-Vautrin J, Mostoslavsky G, Ikonomou L, Kotton DN, Hawkins F, Wilson A, and Villacorta-Martin C
- Abstract
Development of an anti-SARS-CoV-2 therapeutic is hindered by the lack of physiologically relevant model systems that can recapitulate host-viral interactions in human cell types, specifically the epithelium of the lung. Here, we compare induced pluripotent stem cell (iPSC)-derived alveolar and airway epithelial cells to primary lung epithelial cell controls, focusing on expression levels of genes relevant for COVID-19 disease modeling. iPSC-derived alveolar epithelial type II-like cells (iAT2s) and iPSC-derived airway epithelial lineages express key transcripts associated with lung identity in the majority of cells produced in culture. They express ACE2 and TMPRSS2 , transcripts encoding essential host factors required for SARS-CoV-2 infection, in a minor subset of each cell sub-lineage, similar to frequencies observed in primary cells. In order to prepare human culture systems that are amenable to modeling viral infection of both the proximal and distal lung epithelium, we adapt iPSC-derived alveolar and airway epithelial cells to two-dimensional air-liquid interface cultures. These engineered human lung cell systems represent sharable, physiologically relevant platforms for SARS-CoV-2 infection modeling and may therefore expedite the development of an effective pharmacologic intervention for COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
39. PGD2/DP2 receptor activation promotes severe viral bronchiolitis by suppressing IFN- λ production.
- Author
-
Werder RB, Lynch JP, Simpson JC, Zhang V, Hodge NH, Poh M, Forbes-Blom E, Kulis C, Smythe ML, Upham JW, Spann K, Everard ML, and Phipps S
- Subjects
- Allergens, Animals, Animals, Newborn, Antiviral Agents metabolism, Epithelial Cells pathology, Epithelial Cells virology, Humans, Immunity, Infant, Inflammation pathology, Inflammation virology, Intramolecular Oxidoreductases metabolism, Lung pathology, Lung virology, Mice, Inbred BALB C, Murine pneumonia virus, Receptors, Immunologic antagonists & inhibitors, Receptors, Prostaglandin antagonists & inhibitors, Respiratory Syncytial Virus Infections virology, Respiratory Syncytial Viruses physiology, Up-Regulation, Bronchiolitis, Viral metabolism, Bronchiolitis, Viral pathology, Interferon-gamma biosynthesis, Prostaglandin D2 metabolism, Receptors, Immunologic metabolism, Receptors, Prostaglandin metabolism
- Abstract
Prostaglandin D2 (PGD2) signals through PGD2 receptor 2 (DP2, also known as CRTH2) on type 2 effector cells to promote asthma pathogenesis; however, little is known about its role during respiratory syncytial virus (RSV) bronchiolitis, a major risk factor for asthma development. We show that RSV infection up-regulated hematopoietic prostaglandin D synthase expression and increased PGD2 release by cultured human primary airway epithelial cells (AECs). Moreover, PGD2 production was elevated in nasopharyngeal samples from young infants hospitalized with RSV bronchiolitis compared to healthy controls. In a neonatal mouse model of severe viral bronchiolitis, DP2 antagonism decreased viral load, immunopathology, and morbidity and ablated the predisposition for subsequent asthma onset in later life. This protective response was abolished upon dual DP1/DP2 antagonism and replicated with a specific DP1 agonist. Rather than mediating an effect via type 2 inflammation, the beneficial effects of DP2 blockade or DP1 agonism were associated with increased interferon-λ (IFN-λ) [interleukin-28A/B (IL-28A/B)] expression and were lost upon IL-28A neutralization. In RSV-infected AEC cultures, DP1 activation up-regulated IFN-λ production, which, in turn, increased IFN-stimulated gene expression, accelerating viral clearance. Our findings suggest that DP2 antagonists or DP1 agonists may be useful antivirals for the treatment of viral bronchiolitis and possibly as primary preventatives for asthma., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
40. Plasmacytoid dendritic cells protect from viral bronchiolitis and asthma through semaphorin 4a-mediated T reg expansion.
- Author
-
Lynch JP, Werder RB, Loh Z, Sikder MAA, Curren B, Zhang V, Rogers MJ, Lane K, Simpson J, Mazzone SB, Spann K, Hayball J, Diener K, Everard ML, Blyth CC, Forstner C, Dennis PG, Murtaza N, Morrison M, Ó Cuív P, Zhang P, Haque A, Hill GR, Sly PD, Upham JW, and Phipps S
- Subjects
- Animals, Animals, Newborn, Asthma immunology, Bronchiolitis, Viral etiology, Bronchiolitis, Viral immunology, Child, Child, Preschool, Disease Models, Animal, Fatty Acids, Volatile immunology, Fatty Acids, Volatile metabolism, Female, Humans, Interleukin-10 biosynthesis, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microbiota immunology, Receptor, Interferon alpha-beta antagonists & inhibitors, Receptor, Interferon alpha-beta immunology, Respiratory Syncytial Virus Infections complications, Respiratory Syncytial Virus Infections immunology, Semaphorins antagonists & inhibitors, T-Lymphocytes, Regulatory cytology, Asthma prevention & control, Bronchiolitis, Viral prevention & control, Dendritic Cells immunology, Semaphorins immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Respiratory syncytial virus-bronchiolitis is a major independent risk factor for subsequent asthma, but the causal mechanisms remain obscure. We identified that transient plasmacytoid dendritic cell (pDC) depletion during primary Pneumovirus infection alone predisposed to severe bronchiolitis in early life and subsequent asthma in later life after reinfection. pDC depletion ablated interferon production and increased viral load; however, the heightened immunopathology and susceptibility to subsequent asthma stemmed from a failure to expand functional neuropilin-1
+ regulatory T (T reg) cells in the absence of pDC-derived semaphorin 4a (Sema4a). In adult mice, pDC depletion predisposed to severe bronchiolitis only after antibiotic treatment. Consistent with a protective role for the microbiome, treatment of pDC-depleted neonates with the microbial-derived metabolite propionate promoted Sema4a-dependent T reg cell expansion, ameliorating both diseases. In children with viral bronchiolitis, nasal propionate levels were decreased and correlated with an IL-6high /IL-10low microenvironment. We highlight a common but age-related Sema4a-mediated pathway by which pDCs and microbial colonization induce T reg cell expansion to protect against severe bronchiolitis and subsequent asthma., (© 2018 Lynch et al.)- Published
- 2018
- Full Text
- View/download PDF
41. Allergen-encoding bone marrow transfer inactivates allergic T cell responses, alleviating airway inflammation.
- Author
-
Al-Kouba J, Wilkinson AN, Starkey MR, Rudraraju R, Werder RB, Liu X, Law SC, Horvat JC, Brooks JF, Hill GR, Davies JM, Phipps S, Hansbro PM, and Steptoe RJ
- Abstract
Memory Th2 cell responses underlie the development and perpetuation of allergic diseases. Because these states result from immune dysregulation, established Th2 cell responses represent a significant challenge for conventional immunotherapies. New approaches that overcome the detrimental effects of immune dysregulation are required. We tested whether memory Th2 cell responses were silenced using a therapeutic approach where allergen expression in DCs is transferred to sensitized recipients using BM cells as a vector for therapeutic gene transfer. Development of allergen-specific Th2 responses and allergen-induced airway inflammation was blocked by expression of allergen in DCs. Adoptive transfer studies showed that Th2 responses were inactivated by a combination of deletion and induction of T cell unresponsiveness. Transfer of BM encoding allergen expression targeted to DCs terminated, in an allergen-specific manner, Th2 responses in sensitized recipients. Importantly, when preexisting airway inflammation was present, there was effective silencing of Th2 cell responses, airway inflammation was alleviated, and airway hyperreactivity was reversed. The effectiveness of DC-targeted allergen expression to terminate established Th2 responses in sensitized animals indicates that exploiting cell-intrinsic T cell tolerance pathways could lead to development of highly effective immunotherapies.
- Published
- 2017
- Full Text
- View/download PDF
42. The Absence of Interferon-β Promotor Stimulator-1 (IPS-1) Predisposes to Bronchiolitis and Asthma-like Pathology in Response to Pneumoviral Infection in Mice.
- Author
-
Simpson J, Lynch JP, Loh Z, Zhang V, Werder RB, Spann K, and Phipps S
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Asthma genetics, Bronchiolitis genetics, DEAD Box Protein 58 metabolism, Dendritic Cells metabolism, Host-Pathogen Interactions, Interferon-alpha metabolism, Membrane Glycoproteins metabolism, Mice, Inbred C57BL, Mice, Knockout, Pneumovirus Infections genetics, Pneumovirus Infections metabolism, Signal Transduction, Toll-Like Receptor 7 metabolism, Adaptor Proteins, Signal Transducing deficiency, Asthma metabolism, Bronchiolitis metabolism, Murine pneumonia virus physiology, Pneumovirus Infections virology
- Abstract
Respiratory syncytial virus (RSV)-bronchiolitis is a major cause of infant morbidity and mortality and a risk factor for subsequent asthma. We showed previously that toll-like receptor (TLR)7 in plasmacytoid dendritic cells (pDCs) is critical for protection against bronchiolitis and asthma in mice infected with pneumonia virus of mice (PVM), the mouse homolog of RSV. This lack of redundancy was unexpected as interferon-β promotor stimulator-1 (IPS-1) signalling, downstream of RIG-I-like receptor (RLR) and not TLR7 activation, contributes to host defence in hRSV-inoculated adult mice. To further clarify the role of IPS-1 signalling, we inoculated IPS-1
-/- and WT mice with PVM in early-life, and again in later-life, to model the association between bronchiolitis and asthma. IPS-1 deficiency predisposed to severe PVM bronchiolitis, characterised by neutrophilic inflammation and necroptotic airway epithelial cell death, high mobility group box 1 (HMGB1) and IL-33 release, and downstream type-2 inflammation. Secondary infection induced an eosinophilic asthma-like pathophysiology in IPS-1-/- but not WT mice. Mechanistically, we identified that IPS-1 is necessary for pDC recruitment, IFN-α production and viral control. Our findings suggest that TLR7 and RLR signalling work collaboratively to optimally control the host response to pneumovirus infection thereby protecting against viral bronchiolitis and subsequent asthma.- Published
- 2017
- Full Text
- View/download PDF
43. Coinfection with Blood-Stage Plasmodium Promotes Systemic Type I Interferon Production during Pneumovirus Infection but Impairs Inflammation and Viral Control in the Lung.
- Author
-
Edwards CL, Zhang V, Werder RB, Best SE, Sebina I, James KR, Faleiro RJ, de Labastida Rivera F, Amante FH, Engwerda CR, Phipps S, and Haque A
- Subjects
- Animals, Bronchiolitis, Viral virology, Disease Models, Animal, Female, Inflammation immunology, Inflammation parasitology, Inflammation virology, Interferon-beta blood, Interleukin-10 immunology, Lung immunology, Malaria complications, Plasmodium chabaudi, Pneumovirus pathogenicity, Pneumovirus physiology, Pneumovirus Infections complications, Respiratory Syncytial Virus, Human pathogenicity, Viral Load, Weight Loss, Bronchiolitis, Viral immunology, Coinfection, Interferon-beta immunology, Lung virology, Malaria immunology, Pneumovirus immunology, Pneumovirus Infections immunology
- Abstract
Acute lower respiratory tract infections (ALRTI) are the leading cause of global childhood mortality, with human respiratory syncytial virus (hRSV) being a major cause of viral ALRTI in young children worldwide. In sub-Saharan Africa, many young children experience severe illnesses due to hRSV or Plasmodium infection. Although the incidence of malaria in this region has decreased in recent years, there remains a significant opportunity for coinfection. Recent data show that febrile young children infected with Plasmodium are often concurrently infected with respiratory viral pathogens but are less likely to suffer from pneumonia than are non-Plasmodium-infected children. Here, we hypothesized that blood-stage Plasmodium infection modulates pulmonary inflammatory responses to a viral pathogen but does not aid its control in the lung. To test this, we established a novel coinfection model in which mice were simultaneously infected with pneumovirus of mice (PVM) (to model hRSV) and blood-stage Plasmodium chabaudi chabaudi AS (PcAS) parasites. We found that PcAS infection was unaffected by coinfection with PVM. In contrast, PVM-associated weight loss, pulmonary cytokine responses, and immune cell recruitment to the airways were substantially reduced by coinfection with PcAS. Importantly, PcAS coinfection facilitated greater viral dissemination throughout the lung. Although Plasmodium coinfection induced low levels of systemic interleukin-10 (IL-10), this regulatory cytokine played no role in the modulation of lung inflammation or viral dissemination. Instead, we found that Plasmodium coinfection drove an early systemic beta interferon (IFN-β) response. Therefore, we propose that blood-stage Plasmodium coinfection may exacerbate viral dissemination and impair inflammation in the lung by dysregulating type I IFN-dependent responses to respiratory viruses., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.